Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Infect Chemother ; 26(4): 331-334, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31711831

RESUMO

OBJECTIVE: The Laboratory Risk Indicator for Necrotizing Fasciitis (LRINEC) score is a diagnostic tool for necrotizing soft tissue infection (NSTI), which is validated and is considered to have high diagnostic value. However, some experts criticize LRINEC score for consisting of laboratory test results only. METHODS: In this single-center retrospective study, we created a new scoring system (NSTI assessment score; NAS), which also incorporated vital signs as another diagnostic tool for NSTI using cases from our hospital and also evaluated diagnostic accuracy of LRINEC score. We identified NSTI predictors by comparing 24 NSTI patients and 80 non NSTI patients using uni- and multivariate logistic regression analysis, and created NAS based on odds ratio of variables which are statistically significant in the multivariate model. RESULTS: We identified mean arterial pressure, C-reactive protein, hemoglobin, serum creatinine, and glucose as a predictor for NSTI. The maximum value of NAS was 11 points with the cut-off value of 6. Sensitivity, specificity, positive predictive value, and negative predictive value of the NAS for diagnosis of NSTI were 87.5%, 91.3%, 75.0%, and 96.1%, respectively. Area under the receiver operating characteristic curve was 0.926 (0.851-1.00) for the NAS and 0.903 (0.833-0.973) for the LRINEC score, and they were not statistically different (p = 0.167). CONCLUSION: The NAS has high diagnostic accuracy in predicting NSTI, and is comparable with the LRINEC score. The NAS needs to be validated in other cohorts in the future.


Assuntos
Regras de Decisão Clínica , Fasciite Necrosante/diagnóstico , Infecções dos Tecidos Moles/diagnóstico , Sinais Vitais/fisiologia , Idoso , Estudos de Casos e Controles , Fasciite Necrosante/sangue , Feminino , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Estudos Retrospectivos , Medição de Risco , Infecções dos Tecidos Moles/sangue
2.
J Biol Chem ; 286(21): 19138-48, 2011 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-21471203

RESUMO

Vascular calcification is regulated in a process similar to bone formation. BMP2 (bone morphogenetic protein 2) is essential for osteoblastic differentiation of mesenchymal progenitor cells and thus has been implicated in the development of vascular calcification. Here we examined whether Notch signaling interacts with BMP2 signaling to regulate osteogenic differentiation and mineralization of vascular smooth muscle cells (SMCs). BMP2 alone scarcely induced the expression of alkaline phosphatase (ALP), an ectoenzyme crucially required for active biomineralization, in human aortic SMCs (HASMCs), despite its strong induction in osteoblast precursor MC3T3-E1 cells. Notably, overexpression of the Notch1 intracellular domain (N1-ICD) markedly enhanced BMP2-mediated induction of ALP activity and mineralization of HASMCs. In HASMCs, expression of Msx2 gene, a well documented BMP2 target gene in osteoblasts, was barely induced by BMP2 alone, and N1-ICD clearly enhanced the BMP2-driven Msx2 gene expression. Deletion and site-directed mutation analysis of Msx2 gene promoter revealed that the RBPJk-binding site was necessary for BMP2 responsiveness. Using the RBPJk-deficient cells and siRNA for RBPJk, we showed that RBPJk was required for BMP2 induction of Msx2 gene expression and ALP activity. Moreover, we showed that Smad1, a transcription factor downstream of BMP2 signaling, interacted with N1-ICD to form a complex within the Msx2 promoter. Immunohistochemistry of human calcifying atherosclerotic plaques revealed colocalized expression of Notch1, BMP2, and Msx2. These results indicate that the Notch intracellular domain·RBPJk complex enhances the BMP2-induced Msx2 gene expression by cooperating with Smad1 and suggest that Notch signaling makes vascular SMC responsive to BMP2 and promotes vascular calcification.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Calcinose/metabolismo , Diferenciação Celular , Proteínas de Homeodomínio/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Osteoblastos/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Proteína Morfogenética Óssea 2/genética , Calcinose/genética , Calcinose/patologia , Linhagem Celular , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Osteoblastos/patologia , Receptor Notch1/genética
3.
Int Heart J ; 53(5): 331-5, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23038096

RESUMO

Vascular calcification is an active and regulated process that is similar to bone formation. While calcium channel blockers (CCBs) have been shown to improve outcomes in atherosclerotic vascular disease, it remains unknown whether CCBs have an effect on the process of vascular calcification. Here we investigated whether CCBs inhibit osteogenic differentiation and matrix mineralization of vascular smooth muscle cells induced by Msx2, a key factor of vascular calcification. Human aortic smooth muscle cells (HASMCs) were transduced with adenovirus expressing MSX2 and were treated with 3 distinct CCBs. Azelnidipine, a dihydropyridine subclass of CCBs, significantly decreased alkaline phosphatase (ALP) activity of Msx2-overexpressed HASMCs, whereas verapamil and diltiazem had no effect. Furthermore, azelnidipine, but not verapamil and diltiazem, significantly decreased matrix mineralization of Msx2-overexpressing HASMCs. Azelnidipine significantly attenuated the induction of ALP gene expression by Msx2, a key transcription factor in osteogenesis, while it did not reduce enzymatic activity of ALP. Furthermore, azelnidipine inhibited the ability of Msx2 to activate the ALP gene, but had no effect on Notch-induced Msx2 expression. Given that L-type calcium channels are equally blocked by these CCBs, our results suggest that azelnidipine inhibits the Msx2-dependent process of vascular calcification by mechanisms other than inhibition of calcium channel activity.


Assuntos
Ácido Azetidinocarboxílico/análogos & derivados , Calcificação Fisiológica/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Di-Hidropiridinas/farmacologia , Proteínas de Homeodomínio/antagonistas & inibidores , Músculo Liso Vascular/fisiologia , Osteogênese/efeitos dos fármacos , Fosfatase Alcalina/efeitos dos fármacos , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Ácido Azetidinocarboxílico/farmacologia , Diferenciação Celular , Células Cultivadas , Primers do DNA/química , Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Transfecção , Calcificação Vascular/metabolismo , Calcificação Vascular/fisiopatologia
4.
Acute Med Surg ; 9(1): e790, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36285105

RESUMO

Aim: Continuous intravenous (CIV) infusion of epinephrine for the treatment of anaphylaxis may be required if symptoms do not improve after intramuscular (IM) injection. As CIV infusion permits precise dose adjustment, we compared treatment course and adverse events following CIV infusion and IM injection of epinephrine for the management of anaphylaxis. Methods: Medical records of patients, who were treated for anaphylaxis with epinephrine, were 18 years or older, and were admitted to our department from April 2005 to March 2016, were retrospectively reviewed. The cases were categorized as CIV infusion or IM injection, and treatment course and outcomes were compared between the two groups. Results: Of the 142 eligible cases, there were 78 in the CIV infusion group and 64 in the IM injection group. The CIV infusion group had lower systolic blood pressure, more respiratory symptoms, and higher Acute Physiology and Chronic Health Evaluation II (APACHE II) scores, but required a lower total dose of epinephrine, had fewer adverse events after epinephrine administration, and showed lower incidence of biphasic reactions. In addition, compared with the IM injection group, time to administration of epinephrine was significantly longer (P < 0.001), but time to resolution of symptoms, both from contact and epinephrine administration, was significantly shorter (P < 0.01 and P = 0.03, respectively). Conclusion: Continuous intravenous infusion of epinephrine for the treatment of anaphylaxis may be safe, has fewer adverse events, improves symptoms, and is relatively easy to administer under ready conditions. CIV infusion of epinephrine may also reduce the incidence of biphasic reactions.

5.
Cureus ; 14(1): e21654, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35233326

RESUMO

Antineutrophil cytoplasmic antibody (ANCA)-associated renal-limited vasculitis (RLV) is a minor subtype of small vessel vasculitis characterized by the inflammation of blood vessels, tissue damage, and loss of renal function localized in the kidney without systemic involvements. Here, we report a case of myeloperoxidase (MPO) ANCA-associated RLV in a young adult woman in Japan presenting chronic hematuria and newly overt proteinuria. Percutaneous renal biopsy revealed focal fibro-cellular crescent glomerulonephritis and the absence of other small vasculitides, tubular atrophy, and interstitial fibrosis. Therapeutic intravenous methylprednisolone pulse followed by oral prednisolone was administered as a remission induction. The patient's serum MPO-ANCA level gradually decreased, coinciding with dramatic changes in proteinuria and hematuria after therapeutic glucocorticoid administration. Renal function was maintained within the normal range, and disease activity was well-tolerated throughout the follow-up period for more than 14 weeks. While the incidence of RLV is rare among younger patients, it occurs with asymptomatic hematuria and proteinuria, which is important in differentiating RLV from typical glomerulonephritis. The overall prognosis of ANCA-associated RLV potentially depends on the severity of extrarenal involvements. Early diagnosis, appropriate treatment, and regular maintenance are essential for controlling and treating RLV. Due to the nontypical case presented here, further investigation is recommended to improve the diagnosis strategies and treatment options for this disease.

6.
Am J Respir Cell Mol Biol ; 45(1): 136-44, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21749980

RESUMO

Notch is an ancient cell-signaling system that regulates the specification of cell fate. This study examined the role of Notch in the epithelial-mesenchymal transition (EMT) and myofibroblast differentiation of cultured RLE-6TN cells (i.e., rat alveolar epithelial cells). The activation of Notch, either by ectopic expression of the Notch intracellular domain or by the co-culture of RLE-6TN cells with L-Jagged1 cells, induces the expression of smooth muscle α-actin (SMA) and other mesenchymal marker genes (collagen I and vimentin), and reduces the expression of epithelial marker genes (E-cadherin, occludin, and zonula occludens-1). The pharmacologic inhibition of the endogenous Notch signal significantly inhibited the transforming growth factor-ß (TGF-ß)-induced expression of SMA. Cell migratory capacity was increased by Notch. Luciferase assays revealed that the CC(A/T)(6)GG (CArG) box and the TGF-ß control element (TCE) are required for Notch-induced SMA gene transcription. DNA microarray analysis revealed that members of the TGF-ß family as well as Jagged1 were induced in RLE-6TN cells by Notch. Western blot analysis showed that Notch induced the phosphorylation of Smad3, and the TGF-ß receptor type I/activin receptor-like kinase 5 (ALK5) kinase inhibitor SB431542 markedly reduced the Notch-induced expression of SMA. Enzyme-linked immunosorbent assays confirmed the production of TGF-ß1 from RLE-6TN cells by Notch. Immunohistochemistry of a bleomycin-induced model of pulmonary fibrosis and lung specimens from patients with idiopathic interstitial pneumonias showed that Notch was strongly expressed in myofibroblasts, identified as SMA-positive cells. These data indicate that Notch induces myofibroblast differentiation through a TGF-ß-Smad3 pathway that activates SMA gene transcription in a CArG-dependent and TCE-dependent manner in alveolar epithelial cells. Our data also imply that Notch induces the EMT phenotype, with increased migratory behavior in pulmonary fibrosis.


Assuntos
Fibroblastos/metabolismo , Mioblastos/metabolismo , Alvéolos Pulmonares/metabolismo , Receptores Notch/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Antígenos de Diferenciação/biossíntese , Linhagem Celular , Movimento Celular , Técnicas de Cocultura , Transição Epitelial-Mesenquimal , Fibroblastos/patologia , Regulação da Expressão Gênica , Masculino , Mioblastos/patologia , Fosforilação , Alvéolos Pulmonares/patologia , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Ratos , Ratos Wistar , Elementos de Resposta , Transdução de Sinais
7.
Nat Med ; 8(8): 856-63, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12101409

RESUMO

We recently isolated a Krüppel-like zinc-finger transcription factor 5 (KLF5; also known as BTEB2 and IKLF), which is markedly induced in activated vascular smooth-muscle cells and fibroblasts. Here we describe our analysis of the in vivo function of KLF5 using heterozygous KLF5-knockout mice (Klf5(+/-)). In response to external stress, Klf5(+/-) mice showed diminished levels of arterial-wall thickening, angiogenesis, cardiac hypertrophy and interstitial fibrosis. Also, angiotensin II induced expression of KLF5, which in turn activated platelet-derived growth factor-A (PDGF-A) and transforming growth factor-beta (TGF-beta) expression. In addition, we determined that KLF5 interacted with the retinoic-acid receptor (RAR), that synthetic RAR ligands modulated KLF5 transcriptional activity, and that in vivo administration of RAR ligands affected stress responses in the cardiovascular system in a KLF5-dependent manner. KLF5 thus seems to be a key element linking external stress and cardiovascular remodeling.


Assuntos
Angiotensina II/metabolismo , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Remodelação Ventricular/fisiologia , Angiotensina II/farmacologia , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/metabolismo , Células Cultivadas , Colágeno Tipo IV/metabolismo , Dibenzazepinas/farmacologia , Sistema Digestório/anatomia & histologia , Fenômenos Fisiológicos do Sistema Digestório , Feminino , Genes Reporter , Humanos , Fatores de Transcrição Kruppel-Like , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/citologia , Miocárdio/metabolismo , Transplante de Neoplasias , Neovascularização Fisiológica , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Retinoides/antagonistas & inibidores , Transativadores/genética , Dedos de Zinco
8.
Biochem Biophys Res Commun ; 394(2): 243-8, 2010 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-19903460

RESUMO

Expression of bone-associated proteins and osteoblastic transcription factor Runx2 in arterial cells has been implicated in the development of vascular calcification. However, the signaling upstream of the Runx2-mediated activation of osteoblastic program in vascular smooth muscle cells (VSMC) is poorly understood. We examined the effects of fibroblast growth factor-2 (FGF-2), an important regulator of bone formation, on osteoblastic differentiation of VSMC. Stimulation of cultured rat aortic SMC (RASMC) with FGF-2 induced the expression of the osteoblastic markers osteopontin (OPN) and osteocalcin. Luciferase assays showed that FGF-2 induced osteocyte-specific element (OSE)-dependent transcription. Downregulation of Runx2 by siRNA repressed the basal and FGF-2-stimulated expression of the OPN gene in RASMC. FGF-2 produced hydrogen peroxide in RASMC, as evaluated by fluorescent probe. Induction of OPN expression by FGF-2 was inhibited not only by PD98059 (MEK1 inhibitor) and PP1 (c-Src inhibitor), but also by an antioxidant, N-acetyl cysteine. Nuclear extracts from FGF-2-treated RASMC exhibited increased DNA-binding of Runx2 to its target sequence. Immunohistochemistry of human coronary atherectomy specimens and calcified aortic tissues showed that expression of FGF receptor-1 and Runx2 was colocalized. In conclusion, these results suggest that FGF-2 plays a role in inducing osteoblastic differentiation of VSMC by activating Runx2 through mitogen-activated protein kinase (MAPK)-dependent- and oxidative stress-sensitive-signaling pathways.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Fator 2 de Crescimento de Fibroblastos/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Osteoblastos/metabolismo , Osteogênese , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Proteína Tirosina Quinase CSK , Diferenciação Celular , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Fator 2 de Crescimento de Fibroblastos/farmacologia , Expressão Gênica , Técnicas de Silenciamento de Genes , Marcadores Genéticos , Humanos , Peróxido de Hidrogênio/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Osteopontina/genética , Proteínas Tirosina Quinases/metabolismo , Ratos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Ativação Transcricional , Quinases da Família src
9.
Arterioscler Thromb Vasc Biol ; 29(7): 1104-11, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19407244

RESUMO

OBJECTIVE: Vascular calcification is closely correlated with cardiovascular morbidity and mortality. Here, we demonstrate the role of Notch signaling in osteogenic differentiation and mineralization of vascular smooth muscle cells (SMCs). METHODS AND RESULTS: The Msx2 gene, a key regulator of osteogenesis, was highly induced by coculture with Notch ligand-expressing cells or overexpression of Notch intracellular domains (NICDs) in human aortic SMCs (HASMCs). Furthermore, the Notch1 intracellular domain (N1-ICD) overexpression markedly upregulated alkaline phosphatase (ALP) activity and matrix mineralization of HASMCs. A knockdown experiment with a small interfering RNA confirmed that Msx2 mediated N1-ICD-induced osteogenic conversion of HASMCs. Interestingly, Msx2 induction by N1-ICD was independent of bone morphogenetic protein-2 (BMP-2), an osteogenic morphogen upstream of Msx2. The transcriptional activity of the Msx2 promoter was significantly enhanced by N1-ICD overexpression. The RBP-Jk binding element within the Msx2 promoter was critical to Notch-induced Msx2 gene expression. Correspondingly, N1-ICD overexpression did not induce the Msx2 expression in RBP-Jk-deficient fibroblasts. Immunohistochemistry of human carotid artery specimens revealed localization of Notch1, Jagged1 and Msx2 to fibrocalcific atherosclerotic plaques. CONCLUSIONS: These results imply a new mechanism for osteogenic differentiation of vascular SMCs in which Notch/RBP-Jk signaling directly induces Msx2 gene expression and suggest its crucial role in mediating vascular calcification.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Homeodomínio/genética , Miócitos de Músculo Liso/citologia , Ativação Transcricional , Aorta/citologia , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/fisiologia , Receptores Notch/fisiologia , Transdução de Sinais
10.
Arterioscler Thromb Vasc Biol ; 29(1): 99-106, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18927467

RESUMO

OBJECTIVE: TGFbeta and proliferation/phenotypic switching of smooth muscle cells (SMCs) play a pivotal role in pathogenesis of atherosclerotic and restenotic lesions after angioplasty. We have previously shown that the protein inhibitor of activated STAT (PIAS)1 activates expression of SMC differentiation marker genes including smooth muscle (SM) alpha-actin by interacting with serum response factor (SRF) and class I bHLH proteins. Here, we tested the hypothesis that TGFbeta activates SM alpha-actin through PIAS1. METHODS AND RESULTS: An siRNA specific for PIAS1 and ubc9, an E2-ligase for sumoylation, inhibited TGFbeta-induced expression of SM alpha-actin in cultured SMCs as determined by real-time RT-PCR. Overexpression of PIAS1 increased SM alpha-actin promoter activity in a TGFbeta control element (TCE)-dependent manner. Because the TCE within the SM alpha-actin promoter could mediate repression through interaction with KLF4, we tested whether PIAS1 regulates the function of KLF4 for SMC gene expression. PIAS1 interacted with KLF4 in mammalian two-hybrid and coimmunoprecipitation assays, and overexpression of PIAS1 inhibited KLF4-repression of SM alpha-actin promoter activity. Moreover, PIAS1 promoted degradation of KLF4 through sumoylation. CONCLUSIONS: These results provide evidence that PIAS1 promotes TGFbeta-induced activation of SM alpha-actin gene expression at least in part by promoting sumoylation and degradation of the TCE repressor protein, KLF4.


Assuntos
Actinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Proteínas Inibidoras de STAT Ativados/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Animais , Aorta/fisiologia , Células COS/citologia , Células COS/fisiologia , Chlorocebus aethiops , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Músculo Liso Vascular/fisiologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , RNA/genética , RNA/isolamento & purificação , RNA Interferente Pequeno/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
11.
Clin Case Rep ; 8(12): 3649-3651, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33364020

RESUMO

Subclavian central venous catheterization can cause severe complications, including tension pneumothorax, subcutaneous emphysema, and pneumomediastinum. Re-expansion pulmonary edema after thoracentesis is a life-threatening complication.

12.
J Med Case Rep ; 13(1): 101, 2019 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-31014398

RESUMO

BACKGROUND: Hypophosphatasia is an inherited bone disease characterized by low alkaline phosphatase activity encoded by ALPL. Clinically, hypophosphatasia can be categorized as perinatal, infantile, childhood, and adult forms, as well as odonto-hypophosphatasia, according to the age at first sign or dental manifestations. Adult hypophosphatasia typically presents in middle-aged patients who appear to be in good health in early adulthood and manifests as painful feet caused by recurrent, slow-healing stress fractures of the lower limb. Because the symptoms of adult hypophosphatasia vary and are common, many patients with hypophosphatasia might be not diagnosed accurately and thus may receive inappropriate treatment. CASE PRESENTATION: We report a case of a 35-year-old Japanese woman with low serum alkaline phosphatase detected at a routine medical checkup. She had mild muscle/bone pain but no history of rickets, fractures, or dental problems. Measurement of bone mineral density of the lumbar spine and the femoral neck revealed osteopenia below the expected range for age in a young adult. Abdominal ultrasonography revealed numerous microcalcifications in both kidneys. Analysis of amino acids in urine revealed that phosphoethanolamine was elevated. Low serum alkaline phosphatase activity, elevation of phosphoethanolamine, and low bone mineral density supported the diagnosis of hypophosphatasia. ALPL mutation analysis revealed two mutations: p.Phe327Leu and c.1559delT. These genetic abnormalities were previously reported in perinatal, infantile, and childhood but not adult hypophosphatasia. On the basis of the clinical presentation, laboratory and imaging findings, and genetic analyses, the patient was definitively diagnosed with adult hypophosphatasia. To the best of our knowledge, this is the first case report of adult hypophosphatasia with the compound heterozygous mutations p.Phe327Leu and c.1559delT. CONCLUSIONS: Although the risk of bone fracture was high in this case, treatment approaches differ between osteoporosis and hypophosphatasia. Because adult hypophosphatasia diagnosis is often difficult because of their varied symptoms, hypophosphatasia should be considered in the differential diagnosis of low serum alkaline phosphatase. Early diagnosis is important so that appropriate treatment can be initiated.


Assuntos
Fosfatase Alcalina/sangue , Fraturas Espontâneas/genética , Mutação da Fase de Leitura/genética , Hipofosfatasia/genética , Adulto , Análise Mutacional de DNA , Feminino , Fraturas Espontâneas/sangue , Fraturas Espontâneas/fisiopatologia , Humanos , Hipofosfatasia/sangue , Hipofosfatasia/complicações , Hipofosfatasia/fisiopatologia , Mutação de Sentido Incorreto
13.
Mol Cell Biol ; 25(18): 8009-23, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16135793

RESUMO

Although a critical component of vascular disease is modulation of the differentiated state of vascular smooth muscle cells (SMC), the mechanisms governing SMC differentiation are relatively poorly understood. We have previously shown that E-boxes and the ubiquitously expressed class I basic helix-loop-helix (bHLH) proteins, including E2-2 and E12, are important in regulation of the SMC differentiation marker gene, the SM alpha-actin gene. The aim of the present study was to identify proteins that bind to class I bHLH proteins in SMC and modulate transcriptional regulation of SMC differentiation marker genes. Herein we report that members of the protein inhibitor of activated STAT (PIAS) family interact with class I bHLH factors as well as serum response factor (SRF). PIAS1 interacted with E2-2 and E12 based on yeast two-hybrid screens, mammalian two-hybrid assays, and/or coimmunoprecipitation assays. Overexpression of PIAS1 significantly activated the SM alpha-actin promoter and mRNA expression, as well as SM myosin heavy chain and SM22alpha, whereas a small interfering RNA for PIAS1 decreased activity of these promoters, as well as endogenous mRNA expression, and SRF binding to SM alpha-actin promoter within intact chromatin in cultured SMC. Of significance, PIAS1 bound to SRF and activated SM alpha-actin promoter expression in wild-type but not SRF(-/-) embryonic stem cells. These results provide novel evidence that PIAS1 modulates transcriptional activation of SMC marker genes through cooperative interactions with both SRF and class I bHLH proteins.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas Musculares/genética , Miócitos de Músculo Liso/metabolismo , Fator de Resposta Sérica/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Fatores de Transcrição/metabolismo , Actinas/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Diferenciação Celular/genética , Células Cultivadas , Regulação para Baixo/genética , Expressão Gênica , Marcadores Genéticos/genética , Humanos , Camundongos , Proteínas Inibidoras de STAT Ativados , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ratos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Fatores de Transcrição TCF , Proteína 1 Semelhante ao Fator 7 de Transcrição , Proteína 2 Semelhante ao Fator 7 de Transcrição , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Técnicas do Sistema de Duplo-Híbrido , Proteínas de Peixe-Zebra
14.
Arterioscler Thromb Vasc Biol ; 25(2): 341-7, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15591224

RESUMO

OBJECTIVE: Transforming growth factor-beta1 (TGF-beta1) controls the expression of numerous genes, including smooth muscle cell (SMC)-specific genes and extracellular matrix protein genes. Here we investigated whether c-Src plays a role in TGF-beta1 signaling in mouse embryonic fibroblast C3H10T1/2 cells. METHODS AND RESULTS: TGF-beta1 induction of the SMC contractile protein SM22alpha gene expression was inhibited by PP1 (an inhibitor of Src family kinases) or by C-terminal Src kinase (a negative regulator of c-Src). Induction of SM22alpha by TGF-beta1 was markedly attenuated in SYF cells (c-Src(-), Yes(-), and Fyn(-)) compared with Src(++) cells (c-Src(++), Yes(-), and Fyn(-)). PP1 also inhibited the TGF-beta1-induced expression of serum response factor (SRF), a transcription factor regulating the SMC marker gene expression. Confocal immunofluorescence analysis showed that TGF-beta1 stimulates production of hydrogen peroxide. Antioxidants such as catalase or NAD(P)H oxidase inhibitors such as apocynin inhibited the TGF-beta1-induced expression of SM22alpha. Furthermore, we demonstrate that TGF-beta1 induction of the plasminogen activator inhibitor-1 (PAI-1) gene, which is known to be dependent on Smad but not on SRF, is inhibited by PP1 and apocynin. CONCLUSIONS: Our results suggest that TGF-beta1 activates c-Src and generates hydrogen peroxide through NAD(P)H oxidase, and these signaling pathways lead to the activation of specific sets of genes, including SM22alpha and PAI-1. TGF-beta1 controls the expression of numerous genes, including SM22alpha and PAI-1. We investigated whether c-Src plays a role in TGF-beta1 signaling. TGF-beta1 induction of such genes was significantly reduced in Src family tyrosine kinase-deficient cells, and Csk and pharmacological inhibitors for Src family kinases or antioxidants inhibit the effects of TGF-beta1. These results indicate that c-Src and hydrogen peroxide are required for TGF-beta1 signaling.


Assuntos
Fibroblastos/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Peróxido de Hidrogênio/metabolismo , Proteínas dos Microfilamentos/biossíntese , Proteínas Musculares/biossíntese , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Acetofenonas/farmacologia , Acetilcisteína/farmacologia , Animais , Catalase/farmacologia , Linhagem Celular/efeitos dos fármacos , Linhagem Celular/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Ditiocarb/farmacologia , Fibroblastos/fisiologia , Flavonoides/farmacologia , Fluoresceínas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Marcação de Genes , Camundongos , Camundongos Endogâmicos C3H , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/fisiologia , Naftalenos/farmacologia , Inibidor 1 de Ativador de Plasminogênio/genética , Pirazóis/farmacologia , Pirimidinas/farmacologia , Proteínas Recombinantes de Fusão/fisiologia , Fator de Resposta Sérica/biossíntese , Fator de Resposta Sérica/genética , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3 , Proteína Smad4 , Proteína Smad6 , Sulfonas/farmacologia , Transativadores/genética , Transativadores/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Fator de Crescimento Transformador beta1 , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/deficiência , Quinases da Família src/genética , Quinases da Família src/fisiologia
15.
Arterioscler Thromb Vasc Biol ; 25(11): 2328-34, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16151017

RESUMO

OBJECTIVE: Myocardin is a coactivator of serum response factor (SRF) required for vascular smooth muscle cell (VSMC) differentiation. HERP1 is a transcriptional repressor, which is abundantly expressed in vascular system and is known to function as a target gene of Notch. However, the role of HERP1 in the pathogenesis of vascular lesions remains unknown. The present study characterizes the expression of HERP1 in normal and diseased vessels, and tests the hypothesis that HERP1 inhibits SRF/myocardin-dependent SMC gene expression. METHODS AND RESULTS: Immunohistochemistry revealed that HERP1 and myocardin expression was localized to SMC in the neointima of balloon-injured rat aorta and in human coronary atherosclerotic lesions. Expression of both HERP1 and myocardin was elevated in cultured VSMCs compared with medial SMC. Overexpressed HERP1 inhibited the myocardin-induced SMC marker gene expression in 10T1/2 cells. HERP1 protein interfered with the SRF/CArG-box interaction in vivo and in vitro. Immunoprecipitation assays showed that HERP1 physically interacts with SRF. CONCLUSIONS: HERP1 expression was associated with the SMC proliferation and dedifferentiation in vitro and in vivo. HERP1 may play a role in promoting the phenotypic modulation of VSMCs during vascular injury and atherosclerotic process by interfering with SRF binding to CArG-box through physical association between HERP1 and SRF.


Assuntos
Angioplastia com Balão/efeitos adversos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Doença da Artéria Coronariana/patologia , Músculo Liso Vascular/patologia , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fator de Resposta Sérica/metabolismo , Transativadores/metabolismo , Adulto , Animais , Aorta/lesões , Aorta/patologia , Doenças da Aorta/etiologia , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Aterectomia Coronária , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Células Cultivadas , Doença da Artéria Coronariana/metabolismo , Doença da Artéria Coronariana/terapia , Vasos Coronários/lesões , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Expressão Gênica , Marcadores Genéticos , Humanos , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética , Músculo Liso Vascular/metabolismo , Cadeias Pesadas de Miosina/genética , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/fisiologia , RNA Mensageiro/análise , Ratos , Ratos Wistar , Proteínas Repressoras/genética , Miosinas de Músculo Liso/metabolismo , Transativadores/genética , Túnica Íntima/metabolismo , Túnica Íntima/patologia
17.
Atherosclerosis ; 253: 102-110, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27599364

RESUMO

BACKGROUND AND AIMS: Elevated fibroblast growth factor 23 (FGF23) levels are associated with cardiovascular mortality in patients with chronic kidney disease. However, both clinical and basic research have demonstrated conflicting evidence regarding the pathophysiological role of FGF23 in vascular calcification. The aim of this study was to determine the role of FGF23 in the osteoblastic gene expression in vascular smooth muscle cells (SMCs). METHODS AND RESULTS: We transduce human aortic SMCs (HASMCs) expressing klotho and FGF receptors with the adenovirus expressing human FGF23 (Ad-FGF23). We observed significant decreases in the expression of osteoblast-marker genes including BMP2, BMP4, MSX2, RUNX2 and ALP, as well as reduced calcification. Notably, Ad-FGF23 increased mRNA and protein levels of osteoprotegerin (OPG), and human OPG promoter was activated by FGF23. Moreover, in HASMCs overexpressing klotho, FGF23 upregulated OPG expression, whereas depletion of klotho by siRNA attenuated FGF23-induced OPG expression. Furthermore, in 73 consecutive patients with type 2 diabetes mellitus undergoing cardiac computed tomography to determine coronary calcium scores (CCSs), serum FGF23 levels were positively correlated with OPG independent of phosphate and estimated glomerular filtration rate (eGFR, r = 0.65, p < 0.01). Serum FGF23 levels were significantly elevated in patients with high CCSs (≧100) compared to those with low CCSs (<100). CONCLUSIONS: Our in vitro results indicate that FGF23 suppresses osteoblastic gene expression and induces OPG expression in HASMCs. Together with our cross-sectional clinical assessment, the present study lends support to our hypothesis that FGF23 counteracts osteogenic conversion of vascular SMCs as a part of a compensatory mechanism to mitigate vascular calcification.


Assuntos
Aorta/metabolismo , Fatores de Crescimento de Fibroblastos/sangue , Miócitos de Músculo Liso/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoprotegerina/sangue , Calcinose , Movimento Celular , Proliferação de Células , Células Cultivadas , Estudos Transversais , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator de Crescimento de Fibroblastos 23 , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Glucuronidase/metabolismo , Células HeLa , Humanos , Proteínas Klotho , Músculo Liso Vascular/citologia , Osteoblastos/metabolismo , Fosfatos/química , RNA Interferente Pequeno/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Quinases raf/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 24(9): 1596-601, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15231515

RESUMO

OBJECTIVE: Myocardin, a coactivator of serum response factor, has been shown to be required for expression of multiple CArG-dependent smooth muscle cell (SMC) marker genes. The aim of the present study was to determine whether myocardin alone is sufficient to induce SMC lineage in multipotential stem cells as evidenced by activation of the entire SMC differentiation program. METHODS AND RESULTS: Overexpression of myocardin induced only a subset of SMC marker genes, including smooth muscle (SM) alpha-actin, SM-myosin heavy chain (MHC), SM22alpha, calponin, and desmin in A404 SMC precursor cells, whereas expression of smoothelin-B, aortic carboxypeptidase-like protein, and focal adhesion kinase-related nonkinase, whose promoters lack efficacious CArG elements, was not induced. Similar results were obtained in cultured SMCs, 10T1/2 cells, and embryonic stem cells. Moreover, myocardin inappropriately induced expression of skeletal and cardiac CArG-dependent genes in cultured SMCs. Stable overexpression of dominant-negative myocardin in A404 cells resulted in impaired induction of SM alpha-actin and SM-MHC by all trans-retinoic acid but had no effect on induction of smoothelin-B and aortic carboxypeptidase-like protein expression. CONCLUSIONS: Taken together with previous studies, results demonstrate that myocardin is required for the induction of CArG-dependent SMC marker genes but is not sufficient to initiate the complete SMC differentiation program. We examined whether myocardin induces the entire smooth muscle cell (SMC) differentiation program. Results of the present study showed that myocardin knockdown or overexpression affected only a subset of SMC marker genes in multipotential cells, indicating that myocardin is required but not sufficient to induce SMC lineage.


Assuntos
Regulação da Expressão Gênica , Miócitos de Músculo Liso/metabolismo , Proteínas Nucleares/fisiologia , Elemento de Resposta Sérica , Transativadores/fisiologia , Actinas/biossíntese , Actinas/genética , Adenoviridae/genética , Animais , Aorta/citologia , Bovinos , Diferenciação Celular , Células Cultivadas/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Feminino , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Genes Dominantes , Genes Reporter , Vetores Genéticos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/genética , Especificidade de Órgãos , RNA Interferente Pequeno/farmacologia , Ratos , Proteínas Recombinantes de Fusão/fisiologia , Fator de Resposta Sérica/fisiologia , Transativadores/genética , Ativação Transcricional , Transfecção
19.
Arterioscler Thromb Vasc Biol ; 24(8): 1384-90, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15217807

RESUMO

OBJECTIVE: Transforming growth factor-beta1 (TGFbeta1) and fibroblast growth factor (FGF) families play a pivotal role during vascular development and in the pathogenesis of vascular disease. However, the interaction of intracellular signaling evoked by each of these growth factors is not well understood. The present study was undertaken to examine the molecular mechanisms that mediate the effects of TGFbeta1 and basic FGF (bFGF) on smooth muscle cell (SMC) gene expression. METHODS AND RESULTS: TGFbeta1 induction of SMC gene expression, including smooth muscle protein 22-alpha (SM22alpha) and smooth muscle alpha-actin, was examined in the pluripotent 10T1/2 cells. Marked increase in these mRNA levels by TGFbeta1 was inhibited by c-Src-tyrosine kinase inhibitors and protein synthesis inhibitor cycloheximide. Functional studies with deletion and site-directed mutation analysis of the SM22alpha promoter demonstrated that TGFbeta1 activated the SM22alpha promoter through a CC(A/T-rich)6GG (CArG) box, which serves as a serum response factor (SRF)-binding site. TGFbeta1 increased SRF expression through an increase in transcription of the SRF gene. In the presence of bFGF, TGFbeta1 induction of SMC marker gene expression was significantly attenuated. Transient transfection assays showed that bFGF significantly suppressed induction of the SM22alpha promoter-driven luciferase activity by TGFbeta1, whereas bFGF had no effects on the TGFbeta1-mediated increase in SRF expression and SRF:DNA binding activity. Mitogen-activated protein kinase kinase-1 (MEK1) inhibitor PD98059 abrogated the bFGF-mediated suppression of TGFbeta1-induced SMC gene expression. CONCLUSIONS: Our data suggest that bFGF-induced MEK/extracellular signal-regulated kinase signaling plays an antagonistic role in TGFbeta1-induced SMC gene expression through suppression of the SRF function. These data indicate that opposing effects of bFGF and TGFbeta1 on SMC gene expression control the phenotypic plasticity of SMCs.


Assuntos
Fator 2 de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Quinases da Família src/fisiologia , Actinas/biossíntese , Actinas/genética , Animais , Aorta , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/genética , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Cicloeximida/farmacologia , DNA/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/fisiologia , Camundongos , Camundongos Endogâmicos C3H , Proteínas dos Microfilamentos/biossíntese , Proteínas dos Microfilamentos/genética , Proteínas Musculares/biossíntese , Proteínas Musculares/genética , Mutagênese Sítio-Dirigida , Miócitos de Músculo Liso/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Proteínas Recombinantes/farmacologia , Elemento de Resposta Sérica/efeitos dos fármacos , Fator de Resposta Sérica/metabolismo , Fator de Resposta Sérica/farmacologia , Transcrição Gênica/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1 , Quinases da Família src/antagonistas & inibidores , Calponinas
20.
Arterioscler Thromb Vasc Biol ; 24(9): 1602-7, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15242862

RESUMO

OBJECTIVE: Hex (hematopoietically expressed homeobox), a member of homeobox family of transcription factors, has been implicated in the vascular development because of its expression in hemangioblast, a hypothetical stem cell that gives rise to both angioblasts and hematopoietic lineages. In the present study, we examined the role of Hex in the differentiation of vascular smooth muscle cells. METHODS AND RESULTS: We constructed adenovirus expressing Hex, to which we refer to as AxCA/Hex, and transduced murine embryonic fibroblasts, 10T1/2 cells. Northern blot analyses showed that Hex increased the mRNA levels of smooth muscle alpha-actin and SM22alpha but not of calponin and smooth muscle myosin heavy chain. Transient transfection assays showed that Hex activates the transcription from the SM22alpha promoter in a CArG box-dependent manner. Electrophoretic mobility shift assays demonstrate that Hex is not able to bind to CArG box, but binding of serum responsive factor (SRF) to CArG box is enhanced in AxCA/Hex-transduced cells. Recombinant Hex protein produced by in vitro translation system augmented the binding activity of SRF to CArG box. Immunoprecipitation experiments revealed the physical association between Hex and SRF. CONCLUSIONS: Hex induces transcription of the SM22alpha gene by facilitating the interaction between SRF and its cognate binding site in pluripotent embryonic fibroblasts. This study demonstrates that Hex, a hematopoietically expressed homeobox protein, induces transcription of the SM22alpha gene by facilitating the interaction between SRF and its cognate binding site in embryonic fibroblasts. These findings will provide the clue for understanding the mechanisms by which bone marrow-derived SMC precursor cells undergo differentiation.


Assuntos
Actinas/biossíntese , Regulação da Expressão Gênica/fisiologia , Proteínas de Homeodomínio/fisiologia , Proteínas dos Microfilamentos/biossíntese , Proteínas Musculares/biossíntese , Elemento de Resposta Sérica/genética , Fator de Resposta Sérica/farmacologia , Actinas/genética , Animais , Sítios de Ligação , Células Cultivadas/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Fibroblastos/metabolismo , Genes Reporter , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteínas Recombinantes de Fusão/fisiologia , Fator de Resposta Sérica/metabolismo , Fatores de Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA